1
|
Milicic T, Jotic A, Lalic K, Lukic L, Macesic M, Stanarcic Gajovic J, Stoiljkovic M, Milovancevic M, Rafailovic D, Bozovic A, Lalic NM. Insulin Secretion and Insulin Sensitivity Change in Different Stages of Adult-Onset Type 1 Diabetes: A Cross-Sectional Study. J Clin Med 2025; 14:1109. [PMID: 40004639 PMCID: PMC11856604 DOI: 10.3390/jcm14041109] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2025] [Revised: 02/01/2025] [Accepted: 02/03/2025] [Indexed: 02/27/2025] Open
Abstract
Background/Objectives: Previous studies reported impairments in insulin secretion during different stages of type 1 diabetes (T1D), while data regarding insulin sensitivity and immunological changes are still controversial. We analyzed the following: (a) insulin secretion, (b) insulin sensitivity, and (c) pro-inflammatory interleukin-17 (IL-17) levels in peripheral blood in 17 healthy first-degree relatives in stage 1 (FDRs1) (GAD+, IA2+), 34 FDRs in stage 0 (FDRs0) (GAD-, IA2A-), 24 recent-onset T1D (R-T1D) patients in the insulin-requiring state (IRS), 10 in clinical remission (CR), and 18 healthy unrelated controls (HC). Methods: Insulin secretion was evaluated by an IVGTT and a glucagon stimulation test, expressed as a first-phase insulin response (FPIR) and a basal/stimulated C-peptide. Insulin sensitivity was tested by the euglycemic hyperinsulinemic clamp, expressed as an M value. Results: FDRs1 had a lower FPIR than FDRs0 (p < 0.05) and HC (p < 0.001) but higher than RT1D-IRS (p < 0.001) and RT1D-CR (p < 0.01). Moreover, FDRs1 had lower basal/stimulated C-peptide than FDRs0 (p < 0.01/p < 0.05) and HC (p < 0.001/p = 0.001) but higher levels than RT1D-IRS (p < 0.001/p < 0.001). However, the M value was similar among FDRs1, FDRs0, and HC (p = 1.0) but higher than RT1D-IRS (p < 0.001) and RT1D-CR (p < 0.01), while RT1D-IRS and RT1D-CR had lower M than HC (p < 0.001; p < 0.001; respectively). FDRs1 had higher IL-17 than FDRs0 (p < 0.001) and HC (p < 0.05). RT1D-IRS had higher IL-17 than FDRs0 (p < 0.001) and HC (p < 0.001), which was similar to RT1D-CR vs. FDRs0 (p < 0.001) and HC (p < 0.05). Conclusions: Early changes in pre-T1D might involve an initial decline of insulin secretion associated with a pro-inflammatory attack, which does not influence insulin sensitivity, whereas later, insulin sensitivity deterioration seems to be associated with the prominent reduction in insulin secretion.
Collapse
Affiliation(s)
- Tanja Milicic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Aleksandra Jotic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Katarina Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Ljiljana Lukic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Marija Macesic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Jelena Stanarcic Gajovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Milica Stoiljkovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| | - Mina Milovancevic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
| | - Djurdja Rafailovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
| | - Aleksandra Bozovic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
| | - Nebojsa M. Lalic
- Clinic for Endocrinology, Diabetes and Metabolic Diseases, University Clinical Centre of Serbia, Doktora Subotica 13, 11000 Belgrade, Serbia; (A.J.); (K.L.); (L.L.); (M.M.); (J.S.G.); (M.S.); (M.M.); (D.R.); (A.B.); (N.M.L.)
- Faculty of Medicine, University of Belgrade, Doktora Subotica 8, 11000 Belgrade, Serbia
| |
Collapse
|
2
|
Pang H, Huang G, Xie Z, Zhou Z. The role of regulated necrosis in diabetes and its complications. J Mol Med (Berl) 2024; 102:495-505. [PMID: 38393662 DOI: 10.1007/s00109-024-02421-z] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2023] [Revised: 12/21/2023] [Accepted: 01/16/2024] [Indexed: 02/25/2024]
Abstract
Morphologically, cell death can be divided into apoptosis and necrosis. Apoptosis, which is a type of regulated cell death, is well tolerated by the immune system and is responsible for hemostasis and cellular turnover under physiological conditions. In contrast, necrosis is defined as a form of passive cell death that leads to a dramatic inflammatory response (also referred to as necroinflammation) and causes organ dysfunction under pathological conditions. Recently, a novel form of cell death named regulated necrosis (such as necroptosis, pyroptosis, and ferroptosis) was discovered. Distinct from apoptosis, regulated necrosis is modulated by multiple internal or external factors, but meanwhile, it results in inflammation and immune response. Accumulating evidence has indicated that regulated necrosis is associated with multiple diseases, including diabetes. Diabetes is characterized by hyperglycemia caused by insulin deficiency and/or insulin resistance, and long-term high glucose leads to various diabetes-related complications. Here, we summarize the mechanisms of necroptosis, pyroptosis, and ferroptosis, and introduce recent advances in characterizing the associations between these three types of regulated necrosis and diabetes and its complications.
Collapse
Affiliation(s)
- Haipeng Pang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Gan Huang
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China
| | - Zhiguo Xie
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| | - Zhiguang Zhou
- National Clinical Research Center for Metabolic Diseases, Key Laboratory of Diabetes Immunology (Central South University), Ministry of Education, and Department of Metabolism and Endocrinology, The Second Xiangya Hospital of Central South University, Changsha, 410011, Hunan, China.
| |
Collapse
|
3
|
Pollé OG, Delfosse A, Michoux N, Peeters F, Duchêne G, Louis J, Van Nieuwenhuyse B, Clapuyt P, Lysy PA. Pancreas Imaging of Children with Type 1 Diabetes Reveals New Patterns and Correlations with Pancreatic Functions. Pediatr Diabetes 2023; 2023:3295812. [PMID: 40303255 PMCID: PMC12017098 DOI: 10.1155/2023/3295812] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/24/2023] [Revised: 07/09/2023] [Accepted: 08/07/2023] [Indexed: 05/02/2025] Open
Abstract
Objective To perform a longitudinal characterization of the pancreas in patients with new-onset T1D and investigate the correlations between magnetic resonance imaging (MRI) parameters and pancreatic functions during the first year postdiagnosis. Methods Thirty-one pediatric patients with new-onset T1D and 29 retrospective age-, body mass index-, and sex-matched controls were included in the study. Following hypotheses were investigated: (H1) the value of pancreas volume (PV) parameters in T1D and in controls, (H2) the association between MRI parameters and markers of pancreatic functions, (H3) the ability of MRI parameters to predict glucose homeostasis, (H4) the longitudinal evolution of MRI parameters and glucose homeostasis, per-organ (whole pancreas) and per-subregion (head, body, and tail). Results Patients with new-onset T1D demonstrated a significant decrease of PV at diagnosis compared to controls (-45%), with prepubertal patients having increased pancreas atrophy (+25%) (H1). PV parameters were correlated with C-peptide, and trypsinogen (PVTail and PVHead, respectively). Biparametric regression models including MRI parameters predicted pancreas functions during the first year postdiagnosis (H3). Longitudinal evolution of PV parameters at 1 year postdiagnosis was correlated with PV at diagnosis (R = -0.72) but not with markers of glucose homeostasis (H4). Conclusion Our study shows that longitudinal analysis of pancreases of children with T1D using multiparametric MRI improve the understanding of T1D heterogeneity both in the context of its onset and its evolution.
Collapse
Affiliation(s)
- Olivier G. Pollé
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
- Specialized Pediatrics Service, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Antoine Delfosse
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
- Specialized Pediatrics Service, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| | - Nicolas Michoux
- Department of Radiology, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Frank Peeters
- Department of Radiology, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Gaetan Duchêne
- Department of Radiology, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
- MR Applications, General Electric Healthcare, Diegem, Belgium
| | - Jacques Louis
- Division of Pediatric Endocrinology, Department of Pediatrics, Grand Hôpital de Charleroi, Charleroi, Belgium
| | | | - Philippe Clapuyt
- Department of Radiology, Institut de Recherche Expérimentale et Clinique, Cliniques Universitaires Saint Luc, UCLouvain, Brussels, Belgium
| | - Philippe A. Lysy
- Pôle PEDI, Institut de Recherche Expérimentale et Clinique, UCLouvain, Brussels, Belgium
- Specialized Pediatrics Service, Cliniques Universitaires Saint-Luc, Brussels, Belgium
| |
Collapse
|
4
|
Teitelman G. Abnormal Expression of an Insulin Synthesizing Enzyme in Islets of Adult Autoantibody Positive Donors. J Histochem Cytochem 2022; 70:695-706. [PMID: 36341551 PMCID: PMC9660365 DOI: 10.1369/00221554221138368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2022] [Accepted: 10/17/2022] [Indexed: 11/07/2022] Open
Abstract
The observation that the two active forms of proprotein convertase 1/3 (PC1/3) were differentially expressed in beta cells of normal islets raised the possibility that this heterogeneity is lost during type 1 diabetes (T1D) progression. To test this hypothesis, the expression of the convertase was evaluated by confocal microscopy in sections of human pancreas of autoantibody positive (AA+) and T1D donors and compared with that of control. Islets of T1D pancreas were comprised of beta cells expressing either low or high PC1/3 levels and all islets of a pancreatic section contained only one beta cell type. Pancreata of AA+ donors contained either of these two classes of islets intermixed with normal islets comprised of beta cells with heterogeneous PC1/3 expression. This alteration affected the expression of proinsulin and insulin, which in most AA+ and T1D donors were lower than in controls. The present results indicate that the heterogeneity of PC1/3 expression is lost in all beta cells in a subset islets of AA+ donors and in all islets of T1D donors. These findings suggest that the heterogeneity of PC1/3 expression is a biomarker of human beta cell health and that its loss coincides with the initial stages of T1D.
Collapse
Affiliation(s)
- Gladys Teitelman
- Department of Cell Biology, SUNY Downstate Health
Sciences University, Brooklyn, NY
| |
Collapse
|
5
|
Sims EK, Besser RE, Dayan C, Geno Rasmussen C, Greenbaum C, Griffin KJ, Hagopian W, Knip M, Long AE, Martin F, Mathieu C, Rewers M, Steck AK, Wentworth JM, Rich SS, Kordonouri O, Ziegler AG, Herold KC. Screening for Type 1 Diabetes in the General Population: A Status Report and Perspective. Diabetes 2022; 71:610-623. [PMID: 35316839 PMCID: PMC9114719 DOI: 10.2337/dbi20-0054] [Citation(s) in RCA: 92] [Impact Index Per Article: 30.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Accepted: 01/05/2022] [Indexed: 01/10/2023]
Abstract
Most screening programs to identify individuals at risk for type 1 diabetes have targeted relatives of people living with the disease to improve yield and feasibility. However, ∼90% of those who develop type 1 diabetes do not have a family history. Recent successes in disease-modifying therapies to impact the course of early-stage disease have ignited the consideration of the need for and feasibility of population screening to identify those at increased risk. Existing population screening programs rely on genetic or autoantibody screening, and these have yielded significant information about disease progression and approaches for timing for screening in clinical practice. At the March 2021 Type 1 Diabetes TrialNet Steering Committee meeting, a session was held in which ongoing efforts for screening in the general population were discussed. This report reviews the background of these efforts and the details of those programs. Additionally, we present hurdles that need to be addressed for successful implementation of population screening and provide initial recommendations for individuals with positive screens so that standardized guidelines for monitoring and follow-up can be established.
Collapse
Affiliation(s)
- Emily K. Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN
| | - Rachel E.J. Besser
- Department of Paediatrics, National Institute for Health Research Oxford Biomedical Research Centre, John Radcliffe Hospital, Oxford, U.K
- Wellcome Centre for Human Genetics, University of Oxford, Oxford, U.K
| | - Colin Dayan
- Cardiff University School of Medicine, Cardiff, U.K
| | - Cristy Geno Rasmussen
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | | | | | | | - Mikael Knip
- Pediatric Research Center, Children’s Hospital, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
- Research Program for Clinical and Molecular Metabolism, Faculty of Medicine, University of Helsinki, Helsinki, Finland
- Tampere Center for Child Health Research, Tampere University Hospital, Tampere, Finland
| | - Anna E. Long
- Bristol Medical School, University of Bristol, Bristol, U.K
| | | | - Chantal Mathieu
- Department of Endocrinology, UZ Gasthuisberg, KU Leuven, Leuven, Belgium
| | - Marian Rewers
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - Andrea K. Steck
- Barbara Davis Center for Diabetes, School of Medicine, University of Colorado Anschutz Medical Campus, Aurora, CO
| | - John M. Wentworth
- Departments of Diabetes and Endocrinology and Population Health and Immunity, Royal Melbourne Hospital and Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Stephen S. Rich
- Center for Public Health Genomics, University of Virginia, Charlottesville, VA
| | - Olga Kordonouri
- Kinder und Jugendkrankenhaus Auf der Bult, Hannover, Germany
| | - Anette-Gabriele Ziegler
- Institute of Diabetes Research, Helmholtz Zentrum München, German Research Center for Environmental Health, Munich-Neuherberg, Germany
- School of Medicine, Technical University of Munich, Munich, Germany
| | - Kevan C. Herold
- Department of Immunobiology and Department of Internal Medicine, Yale University, New Haven, CT
| | | |
Collapse
|
6
|
Sahin GS, Lee H, Engin F. An accomplice more than a mere victim: The impact of β-cell ER stress on type 1 diabetes pathogenesis. Mol Metab 2021; 54:101365. [PMID: 34728341 PMCID: PMC8606542 DOI: 10.1016/j.molmet.2021.101365] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2021] [Revised: 10/23/2021] [Accepted: 10/26/2021] [Indexed: 12/26/2022] Open
Abstract
BACKGROUND Pancreatic β-cells are the insulin factory of an organism with a mission to regulate glucose homeostasis in the body. Due to their high secretory activity, β-cells rely on a functional and intact endoplasmic reticulum (ER). Perturbations to ER homeostasis and unmitigated stress lead to β-cell dysfunction and death. Type 1 diabetes (T1D) is a chronic inflammatory disease caused by the autoimmune-mediated destruction of β-cells. Although autoimmunity is an essential component of T1D pathogenesis, accumulating evidence suggests an important role of β-cell ER stress and aberrant unfolded protein response (UPR) in disease initiation and progression. SCOPE OF REVIEW In this article, we introduce ER stress and the UPR, review β-cell ER stress in various mouse models, evaluate its involvement in inflammation, and discuss the effects of ER stress on β-cell plasticity and demise, and islet autoimmunity in T1D. We also highlight the relationship of ER stress with other stress response pathways and provide insight into ongoing clinical studies targeting ER stress and the UPR for the prevention or treatment of T1D. MAJOR CONCLUSIONS Evidence from ex vivo studies, in vivo mouse models, and tissue samples from patients suggest that β-cell ER stress and a defective UPR contribute to T1D pathogenesis. Thus, restoration of β-cell ER homeostasis at various stages of disease presents a plausible therapeutic strategy for T1D. Identifying the specific functions and regulation of each UPR sensor in β-cells and uncovering the crosstalk between stressed β-cells and immune cells during T1D progression would provide a better understanding of the molecular mechanisms of disease process, and may reveal novel targets for development of effective therapies for T1D.
Collapse
Affiliation(s)
- Gulcan Semra Sahin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Hugo Lee
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA
| | - Feyza Engin
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53706, USA; Department of Medicine, Division of Endocrinology, Diabetes & Metabolism, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA; Department of Cell & Regenerative Biology, University of Wisconsin-Madison, School of Medicine and Public Health, Madison, WI, 53705, USA.
| |
Collapse
|
7
|
Eizirik DL, Szymczak F, Alvelos MI, Martin F. From Pancreatic β-Cell Gene Networks to Novel Therapies for Type 1 Diabetes. Diabetes 2021; 70:1915-1925. [PMID: 34417266 PMCID: PMC8576417 DOI: 10.2337/dbi20-0046] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/10/2021] [Indexed: 12/14/2022]
Abstract
Completion of the Human Genome Project enabled a novel systems- and network-level understanding of biology, but this remains to be applied for understanding the pathogenesis of type 1 diabetes (T1D). We propose that defining the key gene regulatory networks that drive β-cell dysfunction and death in T1D might enable the design of therapies that target the core disease mechanism, namely, the progressive loss of pancreatic β-cells. Indeed, many successful drugs do not directly target individual disease genes but, rather, modulate the consequences of defective steps, targeting proteins located one or two steps downstream. If we transpose this to the T1D situation, it makes sense to target the pathways that modulate the β-cell responses to the immune assault-in relation to signals that may stimulate the immune response (e.g., HLA class I and chemokine overexpression and/or neoantigen expression) or inhibit the invading immune cells (e.g., PDL1 and HLA-E expression)-instead of targeting only the immune system, as it is usually proposed. Here we discuss the importance of a focus on β-cells in T1D, lessons learned from other autoimmune diseases, the "alternative splicing connection," data mining, and drug repurposing to protect β-cells in T1D and then some of the initial candidates under testing for β-cell protection.
Collapse
Affiliation(s)
- Decio L Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN
- ULB Center for Diabetes Research and Welbio, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Florian Szymczak
- ULB Center for Diabetes Research and Welbio, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Maria Inês Alvelos
- ULB Center for Diabetes Research and Welbio, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | | |
Collapse
|
8
|
Salami F, N.Tamura R, Elding Larsson H, Lernmark Å, Törn C. Complete blood counts with red blood cell determinants associate with reduced beta-cell function in seroconverted Swedish TEDDY children. Endocrinol Diabetes Metab 2021; 4:e00251. [PMID: 34277975 PMCID: PMC8279594 DOI: 10.1002/edm2.251] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Revised: 03/01/2021] [Accepted: 03/22/2021] [Indexed: 12/20/2022] Open
Abstract
OBJECTIVES To investigate whether changes in complete blood count (CBC) in islet autoantibody positive children with increased genetic risk for type 1 diabetes are associated with oral glucose tolerance tests (OGTT) and HbA1c over time. METHODS The Environmental Determinants of Diabetes in the Young (TEDDY) study follows children with increased risk for type 1 diabetes in the United States, Germany, Sweden and Finland. In the current study, 89 Swedish TEDDY children (median age 8.8 years) positive for one or multiple islet autoantibodies were followed up to 5 (median 2.3) years for CBC, OGTT and HbA1c. A statistical mixed effect model was used to investigate the association between CBC and OGTT or HbA1c. RESULTS HbA1c over time increased by the number of autoantibodies (p < .001). Reduction in mean corpuscular haemoglobin (MCH) and mean cell volume (MCV) was both associated with an increase in HbA1c (p < .001). A reduction in red blood cell (RBC) counts (p = .003), haemoglobin (p = .002) and haematocrit (p = .006) levels was associated with increased fasting glucose. Increased red blood cells, haemoglobin, haematocrit and MCH but decreased levels of red blood cell distribution widths (RDW) were all associated with increased fasting insulin. CONCLUSIONS The decrease in RBC indices with increasing HbA1c and the decrease in RBC and its parameters with increasing fasting glucose in seroconverted children may reflect an insidious deterioration in glucose metabolism associated with islet beta-cell autoimmunity.
Collapse
Affiliation(s)
- Falastin Salami
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Roy N.Tamura
- Health Informatics InstituteDepartment of PediatricsUniversity of South FloridaTampaFloridaUSA
| | - Helena Elding Larsson
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Åke Lernmark
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | - Carina Törn
- Department of Clinical SciencesClinical Research CentreLund UniversitySkåne University HospitalMalmöSweden
| | | |
Collapse
|
9
|
Henquin JC. Glucose-induced insulin secretion in isolated human islets: Does it truly reflect β-cell function in vivo? Mol Metab 2021; 48:101212. [PMID: 33737253 PMCID: PMC8065218 DOI: 10.1016/j.molmet.2021.101212] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/02/2021] [Revised: 03/03/2021] [Accepted: 03/09/2021] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Diabetes always involves variable degrees of β-cell demise and malfunction leading to insufficient insulin secretion. Besides clinical investigations, many research projects used rodent islets to study various facets of β-cell pathophysiology. Their important contributions laid the foundations of steadily increasing numbers of experimental studies resorting to isolated human islets. SCOPE OF REVIEW This review, based on an analysis of data published over 60 years of clinical investigations and results of more recent studies in isolated islets, addresses a question of translational nature. Does the information obtained in vitro with human islets fit with our knowledge of insulin secretion in man? The aims are not to discuss specificities of pathways controlling secretion but to compare qualitative and quantitative features of glucose-induced insulin secretion in isolated human islets and in living human subjects. MAJOR CONCLUSIONS Much of the information gathered in vitro can reliably be translated to the in vivo situation. There is a fairly good, though not complete, qualitative and quantitative coherence between insulin secretion rates measured in vivo and in vitro during stimulation with physiological glucose concentrations, but the concordance fades out under extreme conditions. Perplexing discrepancies also exist between insulin secretion in subjects with Type 2 diabetes and their islets studied in vitro, in particular concerning the kinetics. Future projects should ascertain that the experimental conditions are close to physiological and do not alter the function of normal and diabetic islets.
Collapse
Affiliation(s)
- Jean-Claude Henquin
- Unit of Endocrinology and Metabolism, Faculty of Medicine, University of Louvain, Brussels, Belgium.
| |
Collapse
|
10
|
Sims EK, Bundy BN, Stier K, Serti E, Lim N, Long SA, Geyer SM, Moran A, Greenbaum CJ, Evans-Molina C, Herold KC. Teplizumab improves and stabilizes beta cell function in antibody-positive high-risk individuals. Sci Transl Med 2021; 13:eabc8980. [PMID: 33658358 PMCID: PMC8610022 DOI: 10.1126/scitranslmed.abc8980] [Citation(s) in RCA: 191] [Impact Index Per Article: 47.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 02/05/2021] [Indexed: 12/30/2022]
Abstract
We analyzed the effects of a single 14-day course of teplizumab treatment on metabolic function and immune cells among participants in a previously reported randomized controlled trial of nondiabetic relatives at high risk for type 1 diabetes (T1D). In an extended follow-up (923-day median) of a previous report of teplizumab treatment, we found that the median times to diagnosis were 59.6 and 27.1 months for teplizumab- and placebo-treated participants, respectively (HR = 0.457, P = 0.01). Fifty percent of teplizumab-treated but only 22% of the placebo-treated remained diabetes-free. Glucose tolerance, C-peptide area under the curve (AUC), and insulin secretory rates were calculated, and relationships to T cell subsets and function were analyzed. Teplizumab treatment improved beta cell function, reflected by average on-study C-peptide AUC (1.94 versus 1.72 pmol/ml; P = 0.006). Drug treatment reversed a decline in insulin secretion before enrollment, followed by stabilization of the declining C-peptide AUC seen with placebo treatment. Proinsulin:C-peptide ratios after drug treatment were similar between the treatment groups. The changes in C-peptide with teplizumab treatment were associated with increases in partially exhausted memory KLRG1+TIGIT+CD8+ T cells (r = 0.44, P = 0.014) that showed reduced secretion of IFNγ and TNFα. A single course of teplizumab had lasting effects on delay of T1D diagnosis and improved beta cell function in high-risk individuals. Changes in CD8+ T cell subsets indicated that partially exhausted effector cells were associated with clinical response. Thus, this trial showed improvement in metabolic responses and delay of diabetes with immune therapy.
Collapse
Affiliation(s)
- Emily K Sims
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Brian N Bundy
- Department of Epidemiology, and Pediatrics University of South Florida, Tampa, FL 33612, USA
| | - Kenneth Stier
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA
| | | | - Noha Lim
- Immune Tolerance Network, Bethesda, MD 20814, USA
| | - S Alice Long
- Benaroya Research Institute, Seattle WA 98101, USA
| | | | - Antoinette Moran
- Department of Pediatrics, University of Minnesota, Minneapolis, MN 55455, USA
| | | | - Carmella Evans-Molina
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN 46202, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Kevan C Herold
- Departments of Immunobiology and Internal Medicine, Yale University, New Haven, CT 06520, USA.
| |
Collapse
|
11
|
Syed F, Tersey SA, Turatsinze JV, Felton JL, Kang NJ, Nelson JB, Sims EK, Defrance M, Bizet M, Fuks F, Cnop M, Bugliani M, Marchetti P, Ziegler AG, Bonifacio E, Webb-Robertson BJ, Balamurugan AN, Evans-Molina C, Eizirik DL, Mather KJ, Arslanian S, Mirmira RG. Circulating unmethylated CHTOP and INS DNA fragments provide evidence of possible islet cell death in youth with obesity and diabetes. Clin Epigenetics 2020; 12:116. [PMID: 32736653 PMCID: PMC7393900 DOI: 10.1186/s13148-020-00906-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2020] [Accepted: 07/14/2020] [Indexed: 12/30/2022] Open
Abstract
BACKGROUND Identification of islet β cell death prior to the onset of type 1 diabetes (T1D) or type 2 diabetes (T2D) might allow for interventions to protect β cells and reduce diabetes risk. Circulating unmethylated DNA fragments arising from the human INS gene have been proposed as biomarkers of β cell death, but this gene alone may not be sufficiently specific to report β cell death. RESULTS To identify new candidate genes whose CpG sites may show greater specificity for β cells, we performed unbiased DNA methylation analysis using the Infinium HumanMethylation 450 array on 64 human islet preparations and 27 non-islet human tissues. For verification of array results, bisulfite DNA sequencing of human β cells and 11 non-β cell tissues was performed on 5 of the top 10 CpG sites that were found to be differentially methylated. We identified the CHTOP gene as a candidate whose CpGs show a greater frequency of unmethylation in human islets. A digital PCR strategy was used to determine the methylation pattern of CHTOP and INS CpG sites in primary human tissues. Although both INS and CHTOP contained unmethylated CpG sites in non-islet tissues, they occurred in a non-overlapping pattern. Based on Naïve Bayes classifier analysis, the two genes together report 100% specificity for islet damage. Digital PCR was then performed on cell-free DNA from serum from human subjects. Compared to healthy controls (N = 10), differentially methylated CHTOP and INS levels were higher in youth with new onset T1D (N = 43) and, unexpectedly, in healthy autoantibody-negative youth who have first-degree relatives with T1D (N = 23). When tested in lean (N = 32) and obese (N = 118) youth, increased levels of unmethylated INS and CHTOP were observed in obese individuals. CONCLUSION Our data suggest that concurrent measurement of circulating unmethylated INS and CHTOP has the potential to detect islet death in youth at risk for both T1D and T2D. Our data also support the use of multiple parameters to increase the confidence of detecting islet damage in individuals at risk for developing diabetes.
Collapse
Affiliation(s)
- Farooq Syed
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Sarah A Tersey
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, 900 E. 57th Street, KCBD-8130, Chicago, IL, 60637, USA
| | | | - Jamie L Felton
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Nicole Jiyun Kang
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Jennifer B Nelson
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, 900 E. 57th Street, KCBD-8130, Chicago, IL, 60637, USA
| | - Emily K Sims
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Mathieu Defrance
- Laboratory for Cancer Epigenetics, Faculty of Medicine, and ULB Cancer Research Center, Université Libre de Bruxelles, Brussels, Belgium
| | - Martin Bizet
- Laboratory for Cancer Epigenetics, Faculty of Medicine, and ULB Cancer Research Center, Université Libre de Bruxelles, Brussels, Belgium
| | - Francois Fuks
- Laboratory for Cancer Epigenetics, Faculty of Medicine, and ULB Cancer Research Center, Université Libre de Bruxelles, Brussels, Belgium
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Division of Endocrinology (ULB Erasmus Hospital), Université Libre de Bruxelles, Brussels, Belgium
| | - Marco Bugliani
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | - Piero Marchetti
- Department of Clinical and Experimental Medicine, University of Pisa, Pisa, Italy
| | | | | | | | - Appakalai N Balamurugan
- Department of Surgery, Cardiovascular Innovation Institute, University of Louisville, Louisville, KY, USA
- Division of Pediatric General and Thoracic Surgery, Cincinnati Children's Hospital Medical Center, Department of Surgery, University of Cincinnati, Cincinnati, OH, 45229, USA
| | - Carmella Evans-Molina
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Pediatrics, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
- Roudebush VA Medical Center, Indianapolis, IN, USA
| | - Decio L Eizirik
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium
- Indiana Biosciences Research Institute, Indianapolis, IN, USA
| | - Kieren J Mather
- Center for Diabetes and Metabolic Diseases, Indiana University School of Medicine, Indianapolis, IN, USA
- Department of Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Silva Arslanian
- Children's Hospital of Pittsburgh, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - Raghavendra G Mirmira
- Kovler Diabetes Center and Department of Medicine, The University of Chicago, 900 E. 57th Street, KCBD-8130, Chicago, IL, 60637, USA.
| |
Collapse
|
12
|
Eizirik DL, Pasquali L, Cnop M. Pancreatic β-cells in type 1 and type 2 diabetes mellitus: different pathways to failure. Nat Rev Endocrinol 2020; 16:349-362. [PMID: 32398822 DOI: 10.1038/s41574-020-0355-7] [Citation(s) in RCA: 486] [Impact Index Per Article: 97.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/24/2020] [Indexed: 12/12/2022]
Abstract
Loss of functional β-cell mass is the key mechanism leading to the two main forms of diabetes mellitus - type 1 diabetes mellitus (T1DM) and type 2 diabetes mellitus (T2DM). Understanding the mechanisms behind β-cell failure is critical to prevent or revert disease. Basic pathogenic differences exist in the two forms of diabetes mellitus; T1DM is immune mediated and T2DM is mediated by metabolic mechanisms. These mechanisms differentially affect early β-cell dysfunction and eventual fate. Over the past decade, major advances have been made in the field, mostly delivered by studies on β-cells in human disease. These advances include studies of islet morphology and human β-cell gene expression in T1DM and T2DM, the identification and characterization of the role of T1DM and T2DM candidate genes at the β-cell level and the endoplasmic reticulum stress signalling that contributes to β-cell failure in T1DM (mostly IRE1 driven) and T2DM (mostly PERK-eIF2α dependent). Here, we review these new findings, focusing on studies performed on human β-cells or on samples obtained from patients with diabetes mellitus.
Collapse
Affiliation(s)
- Décio L Eizirik
- ULB Center for Diabetes Research, Welbio Investigator, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium.
- Indiana Biosciences Research Institute (IBRI), Indianapolis, IN, USA.
| | - Lorenzo Pasquali
- Endocrine Regulatory Genomics, Department of Experimental & Health Sciences, University Pompeu Fabra, Barcelona, Spain.
- Germans Trias i Pujol University Hospital and Research Institute, Badalona, Spain.
- Josep Carreras Leukaemia Research Institute, Barcelona, Spain.
| | - Miriam Cnop
- ULB Center for Diabetes Research, Université Libre de Bruxelles, Brussels, Belgium.
- Division of Endocrinology, Erasmus Hospital, Université Libre de Bruxelles, Brussels, Belgium.
| |
Collapse
|